EP3411517B1 - Procédé d'identification de patients à haut risque souffrant de lam - Google Patents

Procédé d'identification de patients à haut risque souffrant de lam Download PDF

Info

Publication number
EP3411517B1
EP3411517B1 EP17746659.6A EP17746659A EP3411517B1 EP 3411517 B1 EP3411517 B1 EP 3411517B1 EP 17746659 A EP17746659 A EP 17746659A EP 3411517 B1 EP3411517 B1 EP 3411517B1
Authority
EP
European Patent Office
Prior art keywords
aml
genes
lsc17
score
patients
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP17746659.6A
Other languages
German (de)
English (en)
Other versions
EP3411517A4 (fr
EP3411517A1 (fr
Inventor
Jean Wang
John Dick
Stanley Ng
Mark Minden
Amanda MITCHELL
Weihsu Claire CHEN
Peter Zandstra
James Kennedy
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University Health Network
University of Health Network
Original Assignee
University Health Network
University of Health Network
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University Health Network, University of Health Network filed Critical University Health Network
Priority claimed from PCT/CA2017/000025 external-priority patent/WO2017132749A1/fr
Publication of EP3411517A1 publication Critical patent/EP3411517A1/fr
Publication of EP3411517A4 publication Critical patent/EP3411517A4/fr
Application granted granted Critical
Publication of EP3411517B1 publication Critical patent/EP3411517B1/fr
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57426Specifically defined cancers leukemia
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/04Libraries containing only organic compounds
    • C40B40/06Libraries containing nucleotides or polynucleotides, or derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/04Libraries containing only organic compounds
    • C40B40/06Libraries containing nucleotides or polynucleotides, or derivatives thereof
    • C40B40/08Libraries containing RNA or DNA which encodes proteins, e.g. gene libraries
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B25/00ICT specially adapted for hybridisation; ICT specially adapted for gene or protein expression
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B99/00Subject matter not provided for in other groups of this subclass
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/118Prognosis of disease development
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the invention relates to gene-related methods and products for the identification of high-risk acute myeloid leukemia (AML) patients, and methods of predicting response to treatment.
  • AML acute myeloid leukemia
  • cytogenetically normal (CN) patients with NPM1 mutation and without FLT3-ITD are considered better risk, but -35% relapse within 2 years following conventional therapy 6 .
  • patients with cytogenetically- or mutationally-defined adverse risk features do poorly with standard induction therapies, and although patients with non-responding disease can often achieve remission with second-line therapy, a higher number of chemotherapy cycles required to achieve morphologic remission portends a worse outcome, with or without allogeneic stem-cell transplantation (allo-SCT) 7;8 .
  • allo-SCT allogeneic stem-cell transplantation
  • LSC leukemia stem cells
  • LSCs are best assayed by xenotransplantation into immune-deficient mice, as cell surface phenotype alone is unreliable 17;18 .
  • Xenotransplantation assays have been used to elucidate the biologic properties of normal hematopoietic stem cells (HSCs) 19-22 , and there is a growing body of evidence that these assays also detect clinically relevant properties of LSCs 18;23;24 .
  • Patent application WO2005/080601 relates to methods of genetic analysis for the classification, diagnosis and prognosis of acute myeloid leukemia (AML).
  • the invention provides a method for producing a classification scheme for AML comprising the steps of a) providing a plurality of reference samples, said reference samples comprising cell samples from a plurality of reference subjects affected by AML; b) providing reference profiles by establishing a gene expression profile for each of said reference samples individually; c) clustering said individual reference profiles according to similarity, and d) assigning an AML class to each cluster.
  • the invention further relates to a method for classifying the AML of an AML affected subject, to a method for diagnosing AML in a subject, and to a method of determining the prognosis for an AML affected subject.
  • the document does not disclose the methods or genes of the present invention.
  • a method of prognosing or classifying a subject with AML comprising: (a) determining the expression level of at least 3 genes in a test sample from the subject selected from the group consisting of DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, GPR56; and (b) comparing expression of the at least 3 genes in the test sample with reference expression levels of the at least 3 genes from control samples from a cohort of patients; wherein a difference or similarity in the expression of the at least 3 genes in the test sample and the reference expression levels is used to prognose or classify the subject with AML into a low risk group or a high risk group for worse survival.
  • the method further comprises calculating a LSC Score comprising the weighted sum of expression of each of the at least 3 genes.
  • classification of the subject into a high-risk group is based on a high LSC Score in reference to the control cohort of AML patients.
  • a method of selecting a therapy for a subject with AML comprising the steps: (a) classifying the subject with AML into a high risk group or a low risk group according to the methods described herein; and (b) selecting a more aggressive therapy, preferably intensified chemotherapy, for the high risk group or a less aggressive therapy, preferably standard chemotherapy, for the low risk group.
  • composition comprising a plurality of isolated nucleic acid sequences, wherein each isolated nucleic acid sequence hybridizes to: (a) the mRNA of at least 3 genes selected from the group consisting of DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, GPR56; and/or (b) a nucleic acid complementary to a), wherein the composition is used to measure the level of expression of the at least 3 genes.
  • an array comprising, for each of at least 3 genes selected from the group consisting of DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, GPR56, one or more polynucleotide probes complementary and hybridizable thereto.
  • a computer program product for use in conjunction with a computer having a processor and a memory connected to the processor, the computer program product comprising a computer readable storage medium having a computer mechanism encoded thereon, wherein the computer program mechanism may be loaded into the memory of the computer and cause the computer to carry out the method described herein.
  • a computer implemented product for predicting a prognosis or classifying a subject with AML comprising: (a) a means for receiving values corresponding to a subject expression profile in a subject sample; (b) a database comprising a reference expression profile representing a control, wherein the subject expression profile and the reference profile each have at least one value representing the expression level of at least 3 genes selected from the group consisting of DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, GPR56; wherein the computer implemented product compares the reference expression profile to the subject biomarker expression profile, wherein a difference in the expression profiles is used to prognose or classify the subject with AML into a low risk group or a high risk group for worse survival.
  • a computer implemented product for guiding therapy for a subject with AML comprising the computer implemented product described herein, wherein the computer implemented product further recommends levels of therapies based on whether the subject with AML has been classified into the low risk group or the high risk group.
  • a computer readable medium having stored thereon a data structure for storing the computer-implemented product described herein.
  • a computer system comprising (a) a database including records comprising a reference expression profile of at least 3 genes selected from the group consisting of DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, GPR56 for a cohort of patients; (b) a user interface capable of receiving a selection of expression levels of the at least 3 genes for use in comparing to the reference expression profile in the database; (c) an output that displays a prediction of prognosis or therapy wherein a difference in the expression profiles is used to prognose or classify the subject with AML into a low risk group or a high risk group of worse survival.
  • kits comprising reagents for detecting the expression of at least 3 genes selected from the group consisting of DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, GPR56 in a sample.
  • a method of predicting therapy resistance in a subject with AML comprising: (a) determining the expression level of at least 3 genes in a test sample from the subject selected from the group consisting of DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, GPR56; and (b) comparing expression of at least 3 genes in the test sample with reference expression levels of at least 3 genes from control samples from a cohort of patients; wherein a difference or similarity in the expression of at least 3 genes in the test sample and the reference expression levels is used to predict therapy resistance.
  • FIG. 15 LSC13 sub-score prediction of CN-LMR patients across several microarray (Panels A-C), RNA-Seq (Panel D), and NanoString (Panels E and F) cohorts.
  • LSC17 LSC signature genes
  • the LSC17 and LSC3 scores provide rapid and accurate identification of high-risk patients for whom conventional chemotherapy is non-curative. These scores will enable evaluation in clinical trials of whether such patients may benefit from novel and/or more intensified therapies during induction or in the post-remission setting.
  • a method of prognosing or classifying a subject with AML comprising: (a) determining the expression level of at least 3 genes in a test sample from the subject selected from the group consisting of DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, GPR56; and (b) comparing expression of the at least 3 genes in the test sample with reference expression levels of the at least 3 genes from control samples from a cohort of patients; wherein a difference or similarity in the expression of the at least 3 genes in the test sample and the reference expression levels is used to prognose or classify the subject with AML into a low risk group or a high risk group for worse survival.
  • the at least 3 genes is at least 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16 or 17 genes.
  • prognosis refers to a clinical outcome group such as a worse survival group or a better survival group associated with a disease subtype which is reflected by a reference profile such as a biomarker reference expression profile or reflected by an expression level of the fifteen biomarkers disclosed herein.
  • the prognosis provides an indication of disease progression and includes an indication of likelihood of death due to leukemia.
  • the clinical outcome class includes a better survival group and a worse survival group.
  • prognosing or classifying means predicting or identifying the clinical outcome group that a subject belongs to according to the subject's similarity to a reference profile or biomarker expression level associated with the prognosis.
  • prognosing or classifying comprises a method or process of determining whether an individual with AML has a better or worse survival outcome, or grouping an individual with AML into a better survival group or a worse survival group, or predicting whether or not an individual with AML will respond to therapy.
  • subject refers to any member of the animal kingdom, preferably a human being and most preferably a human being that has AML or that is suspected of having AML.
  • test sample refers to any fluid, cell or tissue sample from a subject which can be assayed for biomarker expression products and/or a reference expression profile, e.g. genes differentially expressed in subjects with AML according to survival outcome.
  • the sample comprises WBCs obtained from peripheral blood (PB) or bone marrow (BM).
  • RNA includes mRNA transcripts, and/or specific spliced or other alternative variants of mRNA, including anti-sense products.
  • RNA product of the biomarker refers to RNA transcripts transcribed from the biomarkers and/or specific spliced or alternative variants.
  • protein it refers to proteins translated from the RNA transcripts transcribed from the biomarkers.
  • protein product of the biomarker refers to proteins translated from RNA products of the biomarkers.
  • level of expression or “expression level” as used herein refers to a measurable level of expression of the products of biomarkers, such as, without limitation, the level of micro-RNA, messenger RNA transcript expressed or of a specific exon or other portion of a transcript, the level of proteins or portions thereof expressed of the biomarkers, the number or presence of DNA polymorphisms of the biomarkers, the enzymatic or other activities of the biomarkers, and the level of specific metabolites.
  • control refers to a specific value or dataset that can be used to prognose or classify the value e.g expression level or reference expression profile obtained from the test sample associated with an outcome class.
  • a dataset may be obtained from samples from a group of subjects known to have AML and better survival outcome or known to have AML and have worse survival outcome or known to have AML and have benefited from chemotherapy (or intensified chemotherapy) or known to have AML and not have benefited from chemotherapy (or intensified chemotherapy).
  • the expression data of the biomarkers in the dataset can be used to create a control value that is used in testing samples from new patients.
  • control is a predetermined value for the set of at least 3 of the 17 biomarkers obtained from AML patients whose biomarker expression values and survival times are known.
  • control is a predetermined reference profile for the set of at least three of the seventeen biomarkers described herein obtained from patients whose survival times are known.
  • differential expression refers to a difference in the level of expression of the biomarkers that can be assayed by measuring the level of expression of the products of the biomarkers, such as the difference in level of mRNA or a portion thereof expressed. In a preferred embodiment, the difference is statistically significant.
  • difference in the level of expression refers to an increase or decrease in the measurable expression level of a given biomarker, for example as measured by the amount of mRNA as compared with the measurable expression level of a given biomarker in a control.
  • better survival refers to an increased chance of survival as compared to patients in the "worse survival” group.
  • the biomarkers of the application can prognose or classify patients into a “better survival group”. These patients are at a lower risk of death from the disease.
  • wase survival refers to an increased risk of death as compared to patients in the "better survival” group.
  • biomarkers or genes of the application can prognose or classify patients into a "worse survival group". These patients are at greater risk of death or adverse reaction from disease, treatment for the disease or other causes.
  • the biomarker reference expression profile comprises a worse survival group. In another embodiment, the biomarker reference expression profile comprises a better survival group.
  • the method further comprises building a subject GE profile from the determined expression of the at least 3 genes.
  • the method may optionally further comprise obtaining a reference GE profile associated with a prognosis, wherein the subject GE profile and the gene reference expression profile each have values representing the expression level of the at least 3 genes.
  • reference expression profile refers to the expression level of at least 3 of the 17 biomarkers selected from DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, and GPR56 associated with a clinical outcome in a AML patient.
  • the reference expression profile comprises 17 values, each value representing the level of a biomarker, wherein each biomarker corresponds to one gene selected from DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, and GPR56.
  • the reference expression profile is identified using one or more samples comprising tumor or adjacent or other-wise tumour-related stromal/blood based tissue or cells, wherein the expression is similar between related samples defining an outcome class or group such as worse survival or better survival and is different to unrelated samples defining a different outcome class such that the reference expression profile is associated with a particular clinical outcome.
  • the reference expression profile is accordingly a reference profile or reference signature of the expression of at least 3 of the 17 biomarkers selected from DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, and GPR56, to which the subject expression levels of the corresponding genes in a patient sample are compared in methods for determining or predicting clinical outcome.
  • the 17 biomarkers selected from DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, and GPR56, to which the subject expression levels of the corresponding genes in a patient sample are compared in methods for determining or predicting clinical outcome
  • the method further comprises calculating a LSC Score comprising the weighted sum expression of each of the at least 3 genes.
  • classification of the subject into a high-risk group is based on a high LSC Score in reference to the control cohort of AML patients. Therefore, in this embodiment the reference expression profile is the expression profile of a reference cohort of AML patients.
  • determination of the LSC17 score is as follows.
  • PB or BM samples are assessed for expression of the 17 signature genes on custom designed NanoString cartridges.
  • each cartridge can hold up to 12 samples to be analyzed.
  • 11 patient RNA samples + 1 control sample comprising a predefined amount of 26 synthetically created oligonucleotides mixed in equal proportions. This includes 17 distinct synthetic oligonucleotides, one created for each of the 17 signature probes + 9 others corresponding to specially selected reference genes that cover a large range of expression of the 17 signature genes.
  • This control is used for reducing non-biological (technical) sources of GE variation that maybe introduced during different cartridge runs.
  • the transcript counts of each control lane on each cartridge are normalized such that they and the RNA counts from the other 11 RNA lanes are comparable across all cartridges. In this way, additional cartridges containing new AML samples that are run at a later time point can be normalized in this way such that new transcript counts are comparable to that of the reference cohort. Ensuring that the expression of the 17 signature genes is comparable across cartridges (new and old) also ensures that LSC17 scores computed from the transcript count data are comparable as well.
  • a threshold value in the reference cohort scores (e.g., median LSC17 score) below which, new scores are considered low and above which are considered high is one possible way to classify patients into low or high-risk categories respectively.
  • the reference dataset and scores do not require much storage and can be distributed with ease in the case of multi-centre studies.
  • the threshold between high and low score would be set based on the distribution of scores in this reference dataset, but future samples need only be run with the reference oligonucleotides in order to be "normalized" to the reference set, and the score calculated. As such, in preferred embodiments, the reference set does not need to be run again.
  • the 9 reference genes are EIF4H, HNRNPK, HNRNPL, PSMA1, PSMD6, SF3B2, SLC25A3, UBE2I, and VPS4A.
  • there are an additional 3 reference genes e.g. ABL1, TBP, and GAPDH).
  • determining the GE level comprises use of quantitative PCR or an array.
  • determining the GE level comprises use of nanostring.
  • biomarkers may be measured using one or more methods and/or tools, including for example, but not limited to, Taqman (Life Technologies, Carlsbad, Calif.), Light-Cycler (Roche Applied Science, Penzberg, Germany), ABI fluidic card (Life Technologies), NanoString.RTM. (NanoString Technologies, Seattle, Wash. and as described in U.S. Pat. No. 7,473,767 ), NANODROP.RTM.
  • nanostring specifically, it is also known to use synthetic oligonucleotides as a control in each nanostring cartridge to minimize inter-cartridge batch effects between runs.
  • the AML is CN-LMR and the at least 3 genes are DPYSL3, AKR1C3, and NYNRIN.
  • a method of selecting a therapy for a subject with AML comprising the steps: (a) classifying the subject with AML into a high risk group or a low risk group according to the methods described herein; and (b) selecting a more aggressive therapy, preferably intensified chemotherapy, for the high risk group or a less aggressive therapy, preferably standard chemotherapy, for the low risk group.
  • Intensified chemotherapy may comprise any chemotherapy that is increased along at least one axis (e.g. dose, duration, frequency, ...etc.) as compared to standard chemotherapy treatment for a particular cancer type and stage.
  • composition comprising a plurality of isolated nucleic acid sequences, wherein each isolated nucleic acid sequence hybridizes to: (a) the mRNA of at least 3 genes selected from the group consisting of DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, GPR56; and/or (b) a nucleic acid complementary to a), wherein the composition is used to measure the level of expression of the at least 3 genes.
  • nucleic acid includes DNA and RNA and can be either double stranded or single stranded.
  • hybridize or “hybridizable” refers to the sequence specific non-covalent binding interaction with a complementary nucleic acid.
  • the hybridization is under high stringency conditions. Appropriate stringency conditions which promote hybridization are known to those skilled in the art, or can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1 6.3.6 . For example, 6.0 x sodium chloride/sodium citrate (SSC) at about 45°C, followed by a wash of 2.0 x SSC at 50°C may be employed.
  • SSC sodium chloride/sodium citrate
  • probe refers to a nucleic acid sequence that will hybridize to a nucleic acid target sequence.
  • the probe hybridizes to the RNA biomarker or a nucleic acid sequence complementary thereof.
  • the length of probe depends on the hybridization conditions and the sequences of the probe and nucleic acid target sequence. In one embodiment, the probe is at least 8, 10, 15, 20, 25, 50, 75, 100, 150, 200, 250, 400, 500 or more nucleotides in length.
  • primer refers to a nucleic acid sequence, whether occurring naturally as in a purified restriction digest or produced synthetically, which is capable of acting as a point of synthesis when placed under conditions in which synthesis of a primer extension product, which is complementary to a nucleic acid strand is induced (e.g. in the presence of nucleotides and an inducing agent such as DNA polymerase and at a suitable temperature and pH).
  • the primer must be sufficiently long to prime the synthesis of the desired extension product in the presence of the inducing agent.
  • the exact length of the primer will depend upon factors, including temperature, sequences of the primer and the methods used.
  • a primer typically contains 15-25 or more nucleotides, although it can contain less or more. The factors involved in determining the appropriate length of primer are readily known to one of ordinary skill in the art.
  • an array comprising, for each of at least 3 genes selected from the group consisting of DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, GPR56, one or more polynucleotide probes complementary and hybridizable thereto.
  • a computer program product for use in conjunction with a computer having a processor and a memory connected to the processor, the computer program product comprising a computer readable storage medium having a computer mechanism encoded thereon, wherein the computer program mechanism may be loaded into the memory of the computer and cause the computer to carry out the method described herein.
  • a computer implemented product for predicting a prognosis or classifying a subject with AML comprising: (a) a means for receiving values corresponding to a subject expression profile in a subject sample; (b) a database comprising a reference expression profile representing a control, wherein the subject expression profile and the reference profile each have at least one value representing the expression level of at least 3 genes selected from the group consisting of DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, GPR56; wherein the computer implemented product compares the reference expression profile to the subject biomarker expression profile, wherein a difference in the expression profiles is used to prognose or classify the subject with AML into a low risk group or a high risk group for poor survival.
  • the computer implemented product calculates a LSC Score comprising the weighted sum expression of each of the at least 3 genes.
  • classification of the subject into a low or high-risk group is based on a high LSC Score in reference to a control cohort of AML patients.
  • a computer implemented product for guiding therapy for a subject with AML comprising the computer implemented product described herein, wherein the computer implemented product further recommends differential levels of therapies based on whether the subject with AML has been classified into the low risk group or the high risk group.
  • a computer readable medium having stored thereon a data structure for storing the computer-implemented product described herein.
  • the data structure is capable of configuring a computer to respond to queries based on records belonging to the data structure, each of the records comprising: (a) a value that identifies a reference expression profile of at least 3 genes selected from the group consisting of DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, GPR56 for a control cohort of patients; (b) a value that identifies the probability of a prognosis associated with the reference expression profile.
  • the value is a LSC Score comprising the weighted sum expression of each of the at least 3 genes.
  • a computer system comprising (a) a database including records comprising a reference expression profile of at least 3 genes selected from the group consisting of DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, GPR56 for a cohort of patients; (b) a user interface capable of receiving a selection of expression levels of the at least 3 genes for use in comparing to the reference expression profile in the database; (c) an output that displays a prediction of prognosis or therapy wherein a difference in the expression profiles is used to prognose or classify the subject with AML into a low risk group or a high risk group of poor survival.
  • kits comprising reagents for detecting the expression of at least 3 genes selected from the group consisting of DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, GPR56 in a sample.
  • a method of predicting therapy resistance in a subject with AML comprising: (a) determining the expression level of at least 3 genes in a test sample from the subject selected from the group consisting of DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3, GPR56; and (b) comparing expression of the at least 3 genes in the test sample with reference expression levels of the at least 3 genes from control samples from a cohort of patients; wherein a difference or similarity in the expression of the at least 3 genes in the test sample and the reference expression levels is used to predict therapy resistance.
  • the at least 3 genes comprise at least 6 genes selected from the group consisting of MMRN1, KIAA0125, CD34, GPR56, LAPTM4B, and NYNRIN.
  • chemotherapy resistance may mean any instance where cancer cells are resisting the effects of the chemotherapy.
  • chemotherapy resistance may occur when cancers that have been responding to a therapy suddenly begin to grow.
  • therapy resistance may be a failure to achieve CR after initial induction.
  • mice 83 samples obtained from 78 AML patients were sorted into fractions based on CD34/CD38 expression prior to transplantation into sublethally irradiated (225 cGy) NSG mice (227 fractions tested). Mice were sacrificed 12 weeks post-transplant and human cell engraftment in the injected right femur (RF) was assessed by flow cytometry using human-specific antibodies: anti-CD3-FITC, anti-CD19-PE, anti-CD33-PE-Cy5, anti-CD45-APC, anti-CD38-PE-Cy7 (all BD Biosciences), and anti-CD34-APC-Cy7 (Beckman Coulter).
  • AML grafts were defined as ⁇ 0.1% human CD45+CD3-cells, with ⁇ 90% CD33 expression. Sorted fractions were defined as LSC+ if transplanted cells generated an AML graft in 1 or more mice; the remaining fractions were defined as LSC-.
  • RNA extracted from 138 LSC+ and 89 LSC- fractions was subjected to GE analysis using Illumina human HT-12 v4 microarrays.
  • RNA extraction was carried out using Qiagen RNeasy mini kits (cat. #74106).
  • Illumina human HT-12 v4 microarrays investigate ⁇ 47,000 targets corresponding to ⁇ 30,000 genes.
  • the resultant fluorescence intensity profiles were subjected to variance stabilization and robust spline normalization using the lumi 2.16.0 R package S4 . All data was put into the log base-2 scale.
  • Differential GE analysis was performed using the limma 3.20.9 package S5 in R. Specifically, Smyth's moderated t-test was used with Benjamini-Hochberg multiple testing correction to compare GE profiles of LSC+ versus LSC-fractions.
  • Raw Affymetrix CEL files were imported using the affy 1.42.3 R package S7 and processed with the gcrma 2.36.0 package S8 in R, which implements the Robust Multiarray Average algorithm that converts background-adjusted probe fluorescence intensities to normalized values in the log 2 scale.
  • version 17 (Released on April 25, 2013) of the custom chip definition files (CDF) S9 for the HGU133 Plus2 platform from the University of Michigan was used.
  • LSC17 score ( DNMT3B ⁇ 0.0874) + ( ZBTB46 ⁇ -0.0347) + ( NYNRIN ⁇ 0.00865) + ( ARHGAP22 ⁇ -0.0138) + ( LAPTM4B ⁇ 0.00582) + ( MMRN1 ⁇ 0.0258) + (DPYSL3 ⁇ 0.0284) + ( KIAA0125 ⁇ 0.0196) + ( CDK6 ⁇ -0.0704) + ( CPXM1 ⁇ -0.0258) + (SOCS2 ⁇ 0.0271) + ( SMIM24 ⁇ -0.0226) + ( EMP1 ⁇ 0.0146) + ( NGFRAP1 ⁇ 0.0465) + ( CD34 ⁇ 0.0338) + ( AKR1C3 ⁇ -0.0402) + ( GPR56 ⁇ 0.0501).
  • LSC17 score ( DNMT3B ⁇ 0.0874) + ( ZBTB46 ⁇ -0.0347) + ( NYNRIN ⁇ 0.0086
  • LSC3 score (DPYSL3 ⁇ 0.3) + ( AKR1C3 ⁇ -0.0477) + ( NYNRIN ⁇ 0.194).
  • a retrained treatment response score was derived by applying the above described regression workflow to a randomly chosen half of the PM cohort.
  • a score comprising 6 of the 17 signature genes resulted: -6.58 + ( MMRN1 ⁇ 0.04) + ( KIAA0125 ⁇ 0.08) + ( CD34 ⁇ 0.10) + ( GPR56 ⁇ 0.20) + ( LAPTM4B ⁇ 0.16) + ( NYNRIN ⁇ 0.12).
  • the LSC17 score was validated against 3 published clinically annotated microarray cohorts with available GE data (2 from GSE12417 S12 , 1 from TCGA S13 , Tables S3-S5), while the LSC3 score was tested on 1 independent CN-LMR cohort (GSE15434 S14 , Table S6). Treatment protocols and the criteria used for cytogenetic/molecular risk classification for each cohort have been previously described S12-14 .
  • Raw Affymetrix CEL files (generated on the HG-U133 A, B, and Plus 2.0 arrays) containing GE data for 2 independent cohorts of CN-AML cases S12 were downloaded from GEO with accession code GSE12417 and clinical annotations were provided by the authors.
  • Raw Affymetrix CEL files (generated on the HG-U133 Plus 2.0 array) containing GE data of a third independent cohort of de novo cytogenetically heterogeneous AML patients S13 were downloaded from TCGA AML data portal (183 cases).
  • An additional dataset of 70 CN-LMR HG-U133 Plus 2.0 array profiles was downloaded from accession GSE15434 to validate the LSC3 CN-LMR specific signature scores, with clinical data provided by the authors S14 . All microarray data were processed as described for the training dataset GSE6891.
  • Signature scores (either LSC17 or LSC3) were determined for each patient in the microarray validation cohorts using the signature gene importance weights derived during training and a median threshold. The scores were then subjected to survival analyses to assess their prognostic value in each cohort.
  • NanoString cartridges implementing this codeset design, each of which comprised 12 lanes.
  • the first 11 lanes of each cartridge were used to measure GE from 11 RNA samples, while the last lane was reserved as a control against which the GE across all cartridges were normalized, in order to minimize inter-cartridge variability as described by others S19;S20 .
  • RNA extraction and quality assessment protocols used in this analysis are described elsewhere S1 .
  • Either 100ng, 150ng, or 250ng of RNA per sample was used to determine the GE levels of the 17 signature and 9 reference genes using the custom NanoString codeset.
  • Total RNA (5 ⁇ L) was incubated with 20 ⁇ L of reporter probe and 5 ⁇ L of capture probe mix (supplied by the manufacturer) at 65°C for 16 to 24 hours for hybridization to take place. The reaction was processed on the nCounter Prep Station (Version 4.0.11.1), while excess probes were washed out using a 2-step magnetic bead-based purification strategy. First, magnetic beads with short nucleic acid sequences complementary to the capture probes are bound to the target/probe complexes in the hybridization mixture.
  • reporter probes and non-target transcripts are then removed.
  • the capture probes and target/probe complexes are eluted off the beads and are re-bound to other magnetic beads with nucleic acid sequences that are complementary to reporter probes.
  • Excess unbound capture probes are removed from the mixture before the purified target/probe complexes are eluted off the beads and immobilized on the NanoString cartridge for data collection.
  • Transcript counts were determined using the nCounter Digital Analyzer (Version 2.1.2.3) at the high-resolution setting. Specifically, digital images were processed with final barcode counts tabulated in reporter code count (RCC) output files containing comma-separated values (CSV).
  • the NanoString assay was performed on 307 diagnostic AML samples from the PM Cancer Centre Tissue Bank collected from patients treated with curative intent between 1999 and 2012 (Table S7). Patients were excluded if they received any cytoreductive treatment other than hydroxyurea or died within one month of starting therapy.
  • RCC files containing raw transcript counts from each cartridge were analyzed using the nSolver analysis software (version 2.0.72) for quality control (QC) and normalization purposes using default settings for GE analysis. Specifically, RCC files for each cartridge along with a reporter library file, which contains probe annotations for our codeset, were imported into nSolver.
  • the software normalized the captured raw counts to the geometric mean of the 9 reference genes included in our assay and the codeset's internal positive controls, while checking for imaging, binding, positive spike-in, and normalization quality.
  • RNA input correction step was used to adjust the GE counts of each cartridge to the reference amount of 100ng RNA.
  • the control lanes for cartridges 1 to 3 were used as blank lanes to estimate per-probe background noise.
  • all signature and reference probe counts were expressed well above 3 standard deviations over background.
  • CV coefficient of variation
  • MFC maximum fold change
  • multiplicative corrective constants were computed and applied to each batch of control lanes according to the oligonucleotide preparation schedule (i.e., cartridges 4, 5 to 11, 12 to 19, 20 to 27 and 28 to 32 represent the batches of oligonucleotides prepared on separate days). Specifically, the oligonucleotide counts of each batch of control lanes were scaled by a ratio of geometric means between the oligonucleotide counts in each batch and that of all control lanes. We next used the batch-corrected control lanes to minimize inter-cartridge technical variation in RNA counts.
  • the geometric mean of the corrected oligonucleotide counts in the control lane of cartridge 5 was divided by the same summary value corresponding to each of the other cartridges to produce per-cartridge scaling factors.
  • the RNA and oligonucleotide counts of each cartridge were then adjusted using these factors by means of multiplication, thereby minimizing batch induced GE variation.
  • CR was defined as achievement of a morphological leukemia-free state (BM blasts ⁇ 5% and absence of extramedullary disease) and recovery of PB counts (absolute neutrophil count ⁇ 1.0 ⁇ 10 9 /L and platelet count ⁇ 100 ⁇ 10 9 /L) S24 .
  • PB counts absolute neutrophil count ⁇ 1.0 ⁇ 10 9 /L and platelet count ⁇ 100 ⁇ 10 9 /L
  • maintenance therapy consisted of ATRA 45 mg/m 2 /d ⁇ 7 days on alternating weeks ⁇ 9 months.
  • maintenance involved 21 monthly cycles of 6-mercaptopurine 75 mg/m 2 /day orally daily for 21 days and methotrexate 20 mg/m 2 /day orally once weekly; every other cycle included ATRA 45 mg/m 2 /d ⁇ 14 days.
  • allo-SCT was performed in CR1 for high-risk patients, typically those with secondary AML, adverse cytogenetics, or normal karyotype with poor prognostic molecular features ( FLT3 -ITD positive; NPM1 mutation positive/ FLT3 -ITD negative).
  • Prerequisites for allo-SCT included disease remission, age less than 71, a lack of significant comorbidities and good performance status.
  • LSC signatures were trained by regression analysis using microarray data from a published cohort of 495 subjects with de novo AML comprising all cytogenetic risk groups, treated with curative intent (GSE6891 25 ).
  • GSE6891 25 For signature validation we used microarray and RNA-seq data from independent AML cohorts (GSE12417 26 , TCGA 27 , and GSE15434 28 ) and NanoString data from the PM cohort.
  • Each LSC signature gene was assigned a regression coefficient, and a score was calculated for each patient in the validation cohorts as the weighted sum of GE of signature genes.
  • the 17-gene signature (LSC17) was re-trained for CN-LMR patients in the GSE6891 cohort to obtain an optimized re-weighted sub-signature comprising 3 of the LSC genes (LSC3). Scores above or below the median of each validation cohort were classified as high or low, respectively. Survival differences between these high-and low-score groups were estimated using uni- and multi-variate survival analyses based on KM and Cox proportional Hazards (CPH) models.
  • CPH Cox proportional Hazards
  • Predictive-ness of the LSC17 score for therapy resistance was quantified by logistic regression analysis using bootstrap-adjusted AUROC.
  • AML GE profiles were compared to those of stem and progenitor cell types purified from human umbilical cord blood (hUCB) (GSE24759 29 and GSE42414 30 ) using the Perturbation model 31 , which estimates the relative proportions of global GE patterns corresponding to multiple blood cell types composing a heterogeneous mixture.
  • hUCB human umbilical cord blood
  • OS was defined as the time from AML diagnosis until death from any cause or last clinical follow-up.
  • EFS was defined as the time from AML diagnosis until an event (defined as induction failure, relapse or death from any cause) or last follow-up.
  • RFS was defined as the time from the date of achievement of first remission until relapse or death (regardless of cause) or last clinical follow-up S24 .
  • TTR was assessed by cumulative incidence analysis with death as a competing risk, and used Gray's test for subgroup comparisons S25 . All survival analyses were performed using the survival 2.38-1 R package S26 . In the comparison to phenotypic signatures, custom CDFs (described previously) were used to summarize microarray probe expression for each gene, since higher HRs resulted compared to scores computed using array probes per gene with maximum average GE in GSE6891.
  • Each of the functionally-defined 138 LSC+ and 89 LSC- fractions was subjected to Illumina microarray GE analysis. GE values were compared between the LSC+ and LSC- fractions to obtain a list of DE genes. 104 genes exhibited at least a 2-fold expression level difference between the 2 groups with an adjusted P value of ⁇ 0.01 ( Figure 1A , Table S8). We defined an LSC+ reference profile as the average expression levels of these 104 genes in the LSC+ fractions.
  • the LSC17 score retained significant prognostic value in all tested cohorts independent of known predictors of outcome including patient age, WBC count, cytogenetic risk group, type of AML (de novo vs. secondary), and the presence of FLT3-ITD and NPM1 mutations (Tables 1, 2, and S9).
  • the LSC17 score displayed superior prognostic accuracy over other published AML signatures that were derived from GE analysis of cell populations defined phenotypically or by multidimensional mass cytometry 34 , demonstrating the power of approaches using cell fractions validated in stem cell assays 18 (Tables S10 and S11).
  • the LSC17 score is strongly associated with survival post-treatment in multiple unrelated AML cohorts that include patients from the spectrum of currently employed risk categories.
  • a GE-based diagnostic test requires a technology that is reproducible, cost-effective, and has rapid turnaround time.
  • the NanoString platform on which an FDA-approved, commercially available prognostic gene signature assay for breast cancer has been implemented 35 , fulfills these criteria and has a large dynamic range for GE measurement 36 .
  • the LSC17 score not only highlights current unmet needs in the treatment of high-risk AML patients, but also provides a rapid and reliable tool to address these needs that is easily implemented on the NanoString platform and can bring about a change in clinical practice.
  • the LSC17 score is the most powerful predictive and prognostic biomarker currently available for AML, and is the first stem cell-based biomarker developed in this way for any human cancer.
  • a good signature should not be composed of too many genes to reduce the chance of statistical overfitting, (2) a good signature should have a high HR in the training dataset, and (3) a good signature should be derived with a minimal number of training cycles to minimize the chance of generating a signature with spurious associations with patient outcome.
  • LSC17 was derived using one training cycle.
  • the LSC17 score substantially increased predictive accuracy in each model, increasing sensitivity and specificity by more than 8% and 1%, respectively (P ⁇ 0.01) and the AUROC to a range of 81.0-83.9.
  • LSC17 score and WBC count were the most predictive covariates as measured by the Wald Chi-squared statistic; however, in the absence of molecular data (which is often not available prior to therapy initiation), the LSC17 score was by far the most predictive covariate. Similar results were seen with the reweighted treatment response score, although there was insufficient molecular data in the testing half of the PM AML cohort for its inclusion in the model. The predictive value of the LSC3 score could not be assessed due to insufficient data-points in the CN-LMR subset. Overall, these data further strengthen the clinical value of the LSC17 score as a tool to predict therapy resistance in newly diagnosed AML patients.
  • LSC17 score retained independent prognostic value in multivariate CPH models including common clinical factors (data not shown). Additionally, a high LSC17 score was significantly associated with shorter survival regardless of whether patients underwent allo-SCT in both the PM (OS, EFS, RFS) and TCGA (OS) AML cohorts as well as their respective CN-AML subsets (data not shown), with a statistically insignificant interaction term between LSC17 score and allo-SCT in CPH models.
  • Table N4 contains the probe sequences designed for our NanoString-based LSC17 diagnostic assay (used in conjunction with the standard chemistry protocol available through Nanostring), which comprises of 17 LSC signature and 9 of the 12 noted reference genes.
  • Table N4 probes can also be used in conjunction with "Elements" chemistry protocol available from Nanostring, which would be used in conjunction with all 12 of the noted reference genes. We found this probeset of particular use, and found, for example a prior codeset design did not yield similarly effective results on the NanoString platform.
  • LSC17 17-gene signature score
  • the LSC17 score identifies a subset of CN-LMR patients with shorter survival, allowing evaluation of the possible benefits of maintenance therapy or early transplantation in first CR.
  • the LSC17 and LSC3 scores implemented on the NanoString platform, will enable rapid identification of high-risk patients for enrolment into clinical trials to evaluate novel approaches to induction and post-induction therapy in an effort to prevent relapse and increase cure rates.
  • AML is an acute condition that progresses quickly and therefore patients will benefit from earlier risk level determination so that the most appropriate treatment strategy can be decided earlier.
  • most AML patients receive standard induction chemotherapy upfront at diagnosis regardless of risk level because risk stratification has depended on cytogenetic and molecular tests that typically take weeks to return results.
  • a small subset of CN-LMR patients are considered to have favourable outcomes, but -35% relapse within 2 years following conventional therapy.
  • a high LSC3 score identifies CN-LMR patients who have poor outcomes following standard induction therapy, and who might benefit from novel frontline therapy.
  • the LSC3 score can only be employed in the post-remission setting, as CN-LMR patients are only identified following cytogenetic and molecular testing.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Physics & Mathematics (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Pathology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Hematology (AREA)
  • Analytical Chemistry (AREA)
  • Biophysics (AREA)
  • Oncology (AREA)
  • Theoretical Computer Science (AREA)
  • Urology & Nephrology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Microbiology (AREA)
  • Spectroscopy & Molecular Physics (AREA)
  • Biomedical Technology (AREA)
  • Medical Informatics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Hospice & Palliative Care (AREA)
  • Evolutionary Biology (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • Food Science & Technology (AREA)
  • Cell Biology (AREA)
  • General Physics & Mathematics (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Claims (11)

  1. Procédé de pronostic ou de classification d'un sujet présentant une LMA comprenant :
    (a) la détermination du niveau d'expression des 17 gènes dans un échantillon d'essai provenant du sujet, consistant en DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3 et GPR56 ; et
    (b) la comparaison de l'expression des 17 gènes de l'échantillon d'essai avec les niveaux d'expression de référence des 17 gènes provenant d'échantillons témoins d'une cohorte de patients ;
    dans lequel la différence ou la similitude dans l'expression des 17 gènes de l'échantillon d'essai et les niveaux d'expression de référence est utilisée pour pronostiquer ou classifier le sujet ayant une LMA en un groupe à faible risque ou un groupe à risque élevé de survie moins élevée.
  2. Procédé selon la revendication 1, comprenant en outre la construction d'un profil d'expression génique du sujet à partir de l'expression déterminée des 17 gènes.
  3. Procédé selon la revendication 2, comprenant en outre l'obtention d'un profil d'expression génique de référence associé à un pronostic, le profil d'expression génique du sujet et le profil d'expression génique de référence ayant chacun des valeurs représentant le niveau d'expression des 17 gènes.
  4. Procédé selon l'une quelconque des revendications 1 à 3, comprenant en outre le calcul du Score des CSL comprenant l'expression de la somme pondérée de chacun des 17 gènes.
  5. Procédé selon la revendication 4, dans lequel la classification du sujet dans un groupe à risque élevé se fonde sur un Score des CSL élevé par référence à la cohorte témoin des patients ayant une LMA.
  6. Procédé selon l'une quelconque des revendications 1 à 5, dans lequel la détermination du niveau d'expression génique comprend l'utilisation d'une PCR quantitative ou d'un micro-réseau, ou en alternance d'un nanostring.
  7. Procédé de sélection d'une thérapie pour un sujet ayant une LMA, comprenant les étapes :
    (a) classification du sujet ayant une LMA en un groupe à risque élevé ou un groupe à faible risque conformément au procédé selon l'une quelconque des revendications 1 à 6 ; et
    (b) la sélection d'une thérapie plus agressive, de préférence une chimiothérapie intensifiée ou une thérapie de remplacement par recrutement dans un essai clinique portant sur une nouvelle thérapie, pour le groupe à risque élevé, ou d'une thérapie moins agressive, de préférence une chimiothérapie standard, pour le groupe à faible risque.
  8. Produit mis en œuvre par ordinateur pour prédire un pronostic ou une classification d'un sujet ayant une LMA, comprenant :
    (a) un moyen pour recevoir des valeurs correspondant à un profil d'expression du sujet dans un échantillon du sujet ;
    (b) une base de données comprenant un profil d'expression de référence représentant un témoin, le profil d'expression du sujet et le profil de référence ayant chacun au moins une valeur représentant le niveau d'expression des 17 gènes consistant en DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3 et GPR56 ;
    le produit mis en œuvre par ordinateur comparant le profil d'expression de référence et le profil d'expression des biomarqueurs du sujet, une différence dans les profils d'expression étant utilisée pour pronostiquer ou classifier le sujet ayant une LMA, en un groupe à faible risque ou un groupe à risque élevé pour une survie moins élevée.
  9. Produit mis en œuvre par ordinateur selon la revendication 8, le produit mis en œuvre par ordinateur calculant un Score des CSL comprenant l'expression de la somme pondérée de chacun des 17 gènes.
  10. Produit mis en œuvre par ordinateur selon la revendication 9, la classification du sujet dans un groupe à faible risque ou à risque élevé se fondant sur un Score des CSL élevé par référence à une cohorte témoin de patients ayant une LMA.
  11. Produit mis en œuvre par ordinateur selon l'une quelconque des revendications 8 à 10, les 17 gènes consistant en DNMT3B, ZBTB46, NYNRIN, ARHGAP22, LAPTM4B, MMRN1, DPYSL3, KIAA0125, CDK6, CPXM1, SOCS2, SMIM24, EMP1, NGFRAP1, CD34, AKR1C3 et GPR56.
EP17746659.6A 2016-02-06 2017-02-06 Procédé d'identification de patients à haut risque souffrant de lam Active EP3411517B1 (fr)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201661292254P 2016-02-06 2016-02-06
US201661348365P 2016-06-10 2016-06-10
US201662426300P 2016-11-24 2016-11-24
US201662429205P 2016-12-02 2016-12-02
PCT/CA2017/000025 WO2017132749A1 (fr) 2016-02-06 2017-02-06 Procédé d'identification de patients à haut risque souffrant de lam

Publications (3)

Publication Number Publication Date
EP3411517A1 EP3411517A1 (fr) 2018-12-12
EP3411517A4 EP3411517A4 (fr) 2020-01-01
EP3411517B1 true EP3411517B1 (fr) 2021-11-17

Family

ID=64048380

Family Applications (1)

Application Number Title Priority Date Filing Date
EP17746659.6A Active EP3411517B1 (fr) 2016-02-06 2017-02-06 Procédé d'identification de patients à haut risque souffrant de lam

Country Status (2)

Country Link
EP (1) EP3411517B1 (fr)
ES (1) ES2903376T3 (fr)

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101088089A (zh) * 2004-02-23 2007-12-12 鹿特丹伊拉斯姆斯大学医疗中心 通过基因表达图形对急性粒细胞性白血病进行分类、诊断和预后
KR100565698B1 (ko) * 2004-12-29 2006-03-28 디지탈 지노믹스(주) 급성골수성백혈병(aml), b-세포형 급성임파구성백혈병(b-all), t 세포형 급성임파구성백혈병(t-all) 진단용 마커
KR100617467B1 (ko) * 2005-09-27 2006-09-01 디지탈 지노믹스(주) 급성 골수성 백혈병 환자의 항암제 치료 반응성 예측용마커
EP2458014A1 (fr) * 2010-09-07 2012-05-30 Rijksuniversiteit Groningen Marqueurs de pronostic pour leucémie myéloïde aiguë (LMA)

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None *

Also Published As

Publication number Publication date
EP3411517A4 (fr) 2020-01-01
EP3411517A1 (fr) 2018-12-12
ES2903376T3 (es) 2022-04-01

Similar Documents

Publication Publication Date Title
Ng et al. A 17-gene stemness score for rapid determination of risk in acute leukaemia
Cavalli et al. Intertumoral heterogeneity within medulloblastoma subgroups
Bill et al. Mutational landscape and clinical outcome of patients with de novo acute myeloid leukemia and rearrangements involving 11q23/KMT2A
Balgobind et al. Evaluation of gene expression signatures predictive of cytogenetic and molecular subtypes of pediatric acute myeloid leukemia
US11111542B2 (en) Method for identifying high-risk AML patients
Bloomston et al. MicroRNA expression patterns to differentiate pancreatic adenocarcinoma from normal pancreas and chronic pancreatitis
Ouillette et al. Integrated genomic profiling of chronic lymphocytic leukemia identifies subtypes of deletion 13q14
Malumbres et al. Paraffin-based 6-gene model predicts outcome in diffuse large B-cell lymphoma patients treated with R-CHOP
Wu et al. Improved risk stratification in myeloma using a micro RNA‐based classifier
AU2009238613A1 (en) Gene expression profiling based identification of genomic signature of high-risk multiple myeloma and uses thereof
JP2015512630A (ja) 急性骨髄性白血病の診断、予後、及び治療用の方法及び組成物
Kopkova et al. Cerebrospinal fluid microRNAs as diagnostic biomarkers in brain tumors
CN110551819B (zh) 一组卵巢癌预后相关基因的应用
KR20140105836A (ko) 다유전자 바이오마커의 확인
Malinge et al. Development of acute megakaryoblastic leukemia in Down syndrome is associated with sequential epigenetic changes
EP2844769B1 (fr) Signature d'expression génique pour la voie de signalisation il-6/stat3, et son utilisation
Scholtysik et al. Characterization of genomic imbalances in diffuse large B‐cell lymphoma by detailed SNP‐chip analysis
Song et al. CBX8 and CD96 are important prognostic biomarkers of colorectal cancer
US20140329714A1 (en) Stat3 activation as a marker for classification and prognosis of dlbcl patients
Wang et al. A robust blood gene expression-based prognostic model for castration-resistant prostate cancer
Shepshelovich et al. MicroRNA signature is indicative of long term prognosis in diffuse large B-cell lymphoma
Rodriguez et al. Identification of a novel recurrent gain on 20q13 in chronic lymphocytic leukemia by array CGH and gene expression profiling
US20210371937A1 (en) Method for identifying high-risk aml patients
Patel et al. Impact of next generation sequencing results on clinical management in patients with hematological disorders
EP3411517B1 (fr) Procédé d'identification de patients à haut risque souffrant de lam

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20180806

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
RIC1 Information provided on ipc code assigned before grant

Ipc: C12Q 1/6886 20180101ALI20190827BHEP

Ipc: C40B 40/08 20060101ALI20190827BHEP

Ipc: C40B 40/06 20060101AFI20190827BHEP

Ipc: G01N 33/48 20060101ALI20190827BHEP

Ipc: C40B 30/04 20060101ALI20190827BHEP

Ipc: C12Q 1/68 20180101ALI20190827BHEP

A4 Supplementary search report drawn up and despatched

Effective date: 20191204

RIC1 Information provided on ipc code assigned before grant

Ipc: C40B 40/06 20060101AFI20191128BHEP

Ipc: C40B 40/08 20060101ALI20191128BHEP

Ipc: C40B 30/04 20060101ALI20191128BHEP

Ipc: C12Q 1/6886 20180101ALI20191128BHEP

Ipc: C12Q 1/68 20180101ALI20191128BHEP

Ipc: G01N 33/48 20060101ALI20191128BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20200910

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20210611

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602017049457

Country of ref document: DE

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1448127

Country of ref document: AT

Kind code of ref document: T

Effective date: 20211215

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG9D

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20211117

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2903376

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20220401

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1448127

Country of ref document: AT

Kind code of ref document: T

Effective date: 20211117

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220217

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220317

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220317

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220217

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220218

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602017049457

Country of ref document: DE

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20220228

26N No opposition filed

Effective date: 20220818

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220206

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220228

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220206

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220228

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220228

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20230220

Year of fee payment: 7

Ref country code: ES

Payment date: 20230317

Year of fee payment: 7

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20230228

Year of fee payment: 7

Ref country code: GB

Payment date: 20230125

Year of fee payment: 7

Ref country code: DE

Payment date: 20230216

Year of fee payment: 7

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20170206

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20211117